WO2017155741A1 - Quinoline analogs as phosphatidylinositol 3-kinase inhibitors - Google Patents

Quinoline analogs as phosphatidylinositol 3-kinase inhibitors Download PDF

Info

Publication number
WO2017155741A1
WO2017155741A1 PCT/US2017/019970 US2017019970W WO2017155741A1 WO 2017155741 A1 WO2017155741 A1 WO 2017155741A1 US 2017019970 W US2017019970 W US 2017019970W WO 2017155741 A1 WO2017155741 A1 WO 2017155741A1
Authority
WO
WIPO (PCT)
Prior art keywords
pi3k5
pharmaceutically acceptable
solvate
compound
acceptable salt
Prior art date
Application number
PCT/US2017/019970
Other languages
French (fr)
Inventor
Xiaolin Hao
Original Assignee
Zhejiang Vimgreen Pharma Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zhejiang Vimgreen Pharma Ltd. filed Critical Zhejiang Vimgreen Pharma Ltd.
Priority to EP17763752.7A priority Critical patent/EP3400227B1/en
Priority to US16/072,150 priority patent/US10792283B2/en
Priority to CA3013490A priority patent/CA3013490C/en
Priority to KR1020187025588A priority patent/KR102256497B1/en
Priority to ES17763752T priority patent/ES2865424T3/en
Priority to KR1020217014690A priority patent/KR20210059033A/en
Priority to CN201780004233.0A priority patent/CN108290898B/en
Priority to JP2018533215A priority patent/JP7201992B2/en
Publication of WO2017155741A1 publication Critical patent/WO2017155741A1/en
Priority to US17/004,734 priority patent/US11446301B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present disclosure relates generally to quinoline analogs as inhibitors of
  • PI3K phosphatidylinositol 3-kinase activity. More specifically, the invention further relates to the preparation of the disclosed PI3K inhibitor analogs and their use in pharmaceutical compositions for the treatment of various diseases, conditions and disorders related to PI3K activity.
  • PIP3 The class I phosphoinositide 3-kinases (PI3Ks) regulate phosphatidylinositol 4,5- bisphosphate (PIP2) phosphorylation.
  • PIP2 phosphatidylinositol 4,5- bisphosphate
  • PIP3 phosphatidylinositol (3,4,5)-trisphosphate
  • PIP3 plays a key regulatory role in cell survival, signal transduction, control of membrane trafficking and other functions.
  • Class I PI3Ks consist of four kinases further delineated into 2 subclasses.
  • Class 1A PI3Ks consist of three closely related kinases, PBKoc, ⁇ , and ⁇ existing as heterodimers composed of a catalytic subunit (pi 10a, ⁇ or ⁇ ) and one of several regulatory subunits. They generally respond to signaling through receptor tyrosine kinases (RTKs).
  • ⁇ 3 ⁇ single class IB isoform responds mainly to G-protein coupled receptors (GPCRs), and is composed of a pi 10 ⁇ catalytic subunit and one of two distinct regulatory subunits.
  • GPCRs G-protein coupled receptors
  • PBKoc and ⁇ are ubiquitously expressed throughout a wide variety of tissue and organ types.
  • PDKy is found mainly in leukocytes, but also in skeletal muscle, liver, pancreas, and heart (Cantly, C. Science 2002, 1655).
  • the expression pattern of PI3K5 is restricted, to spleen, thymus, and peripheral blood leukocytes (Knight, Z. et al. Cell 2006, 125, 733).
  • PI3K5 has been implicated as a major player in the adaptive immune system due to its expression pattern and evidence accumulated with genetically modified mice.
  • activated PI3K delta syndrome APDS
  • PID primary immunodeficiencies
  • E1021K glutamic acid at residue 1021
  • ⁇ inhibitors can potentially be supplemental to the treatments of B-cell related diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) by the biologies rituximab (Rituxan) and belimumab (Benlysta), as well as primary immunodeficiencies (PID).
  • RA rheumatoid arthritis
  • SLE systemic lupus erythematosus
  • PID primary immunodeficiencies
  • PI3K5 selective inhibitors such as, idelalisib (GS-1101), IPI-145 and AMG 319 have entered the clinic targeting hematological malignancies, but few inhibitors have entered clinical trials for anti-inflammatory treatments (Cushing, T. et al. J Med Chem. 2015, 58, 480).
  • idelalisib has a boxed warning describing toxicities that can be serious and fatal, including liver toxicity. Fatal and/or serious hepatotoxicity occurred in 18% of patients treated with idelalisib monotherapy and 11% of patients treated with idelalisib in combination trials. Elevations in ALT or AST are greater than 5 times the upper limit of what has occurred normally. The liver toxicity may be related to the inhibition and induction of CYP enzymes by idelalisib and its metabolite GS-563117.
  • pi 105 inactivation in mice protects against a broad range of cancers, including non-haematological solid tumors, and that pi 105 inactivation in regulatory T cells unleashes CD8(+) cytotoxic T cells and induces tumor regression.
  • pi 105 inhibitors can break tumor-induced immune tolerance and potentially have wide usage in oncology.
  • PI3K delta inhibitor can have improved in vivo stability to overcome the liability of inhibition or induction of CYP enzymes; an ideal PI3K delta inhibitor can have the potential of combination treatment of malignant tumors with other anti-cancer interventions, such as emerging immunotherapies.
  • the present invention provides novel compounds that are inhibitors of PI3K isoforms with significantly improved profiles.
  • compositions including pharmaceutical compositions that include the compounds are also provided, as are methods of using and making the compounds.
  • the compounds provided herein may find use in treating diseases, disorders, or conditions that are mediated by PI3K isoforms, such as PI3K delta.
  • X is N or CH
  • Each Ri and R 2 is independently H, F, or S0 2 Me.
  • R 3 is F or CI. [013] In one embodiment of formula (A) where X is N, both Ri and R 2 are H, R 3 is 7-F.
  • the compound is of compound (2) in Table 1; or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
  • PI3K-mediated conditions or disorders with a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
  • a method of treatments of inflammatory diseases such as asthma, chronic obstructive pulmonary disease, rheumatoid arthritis, multiple sclerosis, and lupus.
  • Also provided is a method of inhibiting the growth or proliferation of cancer cells comprising contacting the cancer cells with an effective amount of a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate. Also provided is a method for increasing sensitivity of cancer cells to chemotherapy, comprising administering to a patient undergoing chemotherapy with a chemo therapeutic agent and a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate, sufficient to increase the sensitivity of cancer cells to the chemotherapeutic agent.
  • articles of manufacture that include a compound of formula (A), or a pharmaceutically acceptable, or prodrug, or solvate thereof.
  • PI3K inhibitors [019] Provided herein are compounds that function as PI3K inhibitors.
  • X is N or CH
  • Each Ri and R 2 are independently H, F, or S0 2 Me.
  • R 3 is F or CI.
  • R 3 is 7-F.
  • the compound is of compound (2) in Table 1; or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
  • “Pharmaceutically-acceptable salt” means a salt prepared by conventional means, and are well known by those skilled in the art. The “pharmacologically acceptable salts” include basic salts of inorganic and organic acids (Berge et al., J. Pharm. Sci. 1977, 66: 1).
  • a "solvate” is formed by treating a compound in a solvent. Solvates of salts of the compounds of formula (A) are also provided. In the case of treating compounds with water, the solvate is hydrates. Hydrates of the compounds of formula (A) are also provided.
  • a "prodrug” includes any compound that converts into a compound of formula (A), when administered to a subject, e.g., upon metabolic processing of the prodrug.
  • the compounds of formula (A), or pharmaceutically acceptable salt, prodrug, or solvate thereof may be used for treating PI3K mediated diseases or disorders.
  • PI3K delta and gamma isomers may both be inhibited to achieve optimal efficacy.
  • selected compounds of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof have improved properties in at least one of the following parameters: (i) human hepatocyte stability, and (ii) pharmacokinetic profiles (PK) including oral exposure.
  • selected compounds of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof have improved human hepatocyte stability.
  • Human hepatocyte stability in many cases correlates with pharmacokinetics in human better than the corresponding rodent pharmacokinetic studies.
  • selected compounds may have hepatocyte stability of a half-life of greater than 24 hours.
  • treat or “treating” in reference to a disorder means to ameliorate or prevent the disorder or one or more of the biological manifestations of the disorder, to interfere with one or more points in the biological cascade that leads to or is responsible for the disorder, to alleviate one or more of the symptoms or effects associated with the disorder.
  • treatment includes prevention of the disorder, and “prevention” is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a disorder or biological manifestation thereof, or to delay the onset of such disorder or biological manifestation thereof.
  • Subject refers to a human (including adults and children) or other animal. In one embodiment, “patient” refers to a human.
  • safe and effective dose in reference to a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof an amount sufficient to treat the patient's condition but low enough to avoid serious side effects.
  • a safe and effective dose of a compound will vary with the particular compound chosen (e.g. consider the potency, efficacy, and half-life of the compound); the route of administration chosen; the disorder being treated; the severity of the disorder being treated; the age, size, weight, and physical condition of the patient being treated; the medical history of the patient to be treated; the duration of the treatment; the nature of concurrent therapy; the desired therapeutic effect; and like factors.
  • PI3K delta activity refers to a decrease in PI3K delta activity as a direct or indirect response to the presence of a compound of formula (A), or a
  • PI3K delta selective inhibitor generally refers to a compound that inhibits the activity of the PI3K delta isoform more effectively than other isoforms of the PI3K family (e.g., PI3K alpha, beta, or gamma).
  • potencies of compounds as inhibitors of an enzyme activity can be established by determining the concentrations at which each compound inhibits the activity to a predefined extent and then comparing the results.
  • concentrations at which each compound inhibits the activity can be determined by the concentration that inhibits 50% or 90% of the activity in a biochemical assay, which can be accomplished using conventional techniques known in the art, including the techniques describes in the Examples below.
  • PI3K delta is expressed primarily in hematopoietic cells including leukocytes such as T- cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages. Due to its integral role in immune system function, PI3K delta is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases. By inhibiting aberrant proliferation of hematopoietic cells, PI3K delta inhibitors can ameliorate the symptoms and secondary conditions that result from a primary effect such as excessive system or localized levels of leukocytes or lymphocytes.
  • the invention thus provides a method of treating a disorder mediated by inappropriate P13-kinase activity comprising administering a safe and effective dose of a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof
  • PI3K mediated diseases or disorders are selected from the group consisting of respiratory diseases (including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF)); allergic diseases (including allergic rhinitis and atopic dermatitis); autoimmune diseases (including rheumatoid arthritis and multiple sclerosis); inflammatory disorders (including inflammatory bowel disease); hematologic malignancies; solid tumors; neurodegenerative diseases; pancreatitis; kidney diseases;
  • respiratory diseases including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF)
  • allergic diseases including allergic rhinitis and atopic dermatitis
  • autoimmune diseases including r
  • transplantation rejection transplantation rejection; graft rejection; lung injuries
  • the compounds described herein may be used to treat cancers that are mediated by inappropriate PI3K delta activity.
  • the disease is a hematologic malignancy.
  • the disease is lymphoma, such as Burkitt lymphoma, diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL), follicular lymphoma, lymphoplasmacytic lymphoma, Waldenstrom macroglobulinemia, and marginal zone lymphoma.
  • the disorder is multiple myeloma, or leukemia, such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic
  • CLL lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MDS myelodysplastic syndrome
  • MPD myeloproliferative disease
  • CML chronic myeloid leukemia
  • the disease is a solid tumor.
  • the indication is to treat solid tumor with abnormal PI3K delta expression, such as pancreatic cancer, gastric cancer, esophageal cancer, and breast cancer.
  • the compounds alone or with combination of other anti-cancer therapies may be used to treat prostate cancer, bladder cancer, colorectal cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, head and neck cancer, melanoma, neuroendocrine cancers, brain tumors, bone cancer, or soft tissue sarcoma.
  • PI3K mediated diseases or disorders are severe autoimmune disease as asthma, type I diabetes, rheumatoid arthritis, multiple sclerosis, chronic obstructive pulmonary disease (COPD), and lupus.
  • COPD chronic obstructive pulmonary disease
  • a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat cancers or inflammatory disorders.
  • the one or more additional therapeutic agents may be a chemotherapeutic agent, a radiotherapy, a targeted therapy, an immunotherapeutic agent or any current best of care treatment, either as a small molecule or a biologic nature.
  • Targeted therapies include but not limit to an inhibitor to cyclin-dependent kinase (CDK) such as CDK1, CDK2, CDK4/6, CDK7, and CDK9, Janus kinase (JAK) such as JAK1, JAK2 and/or JAK3 , spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), mitogen- activated protein kinase (MEK) such as MEK 1 and MEK2, bromodomain containing protein inhibitor (BRD) such as BRD4, isocitrate dehydrogenase (IDH) such as IDH1, histone deacetylase (HDAC), or any combination thereof.
  • CDK cyclin-dependent kinase
  • JAK1 JAK2 and/or JAK3 Janus kinase
  • SYK spleen tyrosine kinase
  • BTK Bruton
  • Chemotherapeutic agents may be categorized by their mechanism of action into:
  • a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with chemotherapeutic s to sensitize and improve the efficacy of certain chemotherapeutic agents to treat blood or solid tumors.
  • the immunotherapeutic agents include and are not limited to therapeutic antibodies, small molecules and vaccines suitable for treating patients; such as IDOl and TD02 inhibitors, A2A receptor inhibitors, arginase inhibitors, toll-like receptor agonists, chemokine regulators (including CCR and CXCR families), check point blockage antibodies such as antibodies that regulate PD-1, PD-Ll, CTLA-4, OX40-OX40 ligand, LAG3, TIM3, or any combination thereof.
  • IDOl and TD02 inhibitors such as IDOl and TD02 inhibitors, A2A receptor inhibitors, arginase inhibitors, toll-like receptor agonists, chemokine regulators (including CCR and CXCR families), check point blockage antibodies such as antibodies that regulate PD-1, PD-Ll, CTLA-4, OX40-OX40 ligand, LAG3, TIM3, or any combination thereof.
  • Radiotherapy is part of cancer treatment to control or kill malignant cells and commonly applied to the cancerous tumor because of its ability to control cell growth.
  • a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with radiotherapy, to improve the efficacy of radiotherapy to treat blood or solid tumors, or with surgery, chemotherapy, immunotherapy and combination of the four.
  • a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat patients who are substantially refractory to at least one chemotherapy treatment, or in relapse after treatment with chemotherapy.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate salt thereof and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and topical administration, etc.
  • compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and can be prepared in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules.
  • Oral composition may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • the tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin or olive oil.
  • compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier.
  • Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and
  • compositions for topical application include aqueous solutions, suspensions, ointments, creams, gels or spray formulations, e. g., for delivery by aerosol or the like.
  • Such topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like.
  • a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray, atomizer or nebulizer, with or without the use of a suitable propellant.
  • a dry powder either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids
  • compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose.
  • agents which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
  • compositions may be administered in either single or multiple doses.
  • a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate salt thereof can be formulated so as to provide the desired release schedule of the active ingredient based on the therapeutic treatment purpose.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules.
  • these may contain an amount of active ingredient from about 0.1 to 1000 mg, preferably from about 0.1 to 500 mg.
  • a suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods. The daily dose can be administered in one to four doses per day.
  • the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration drops suitable for administration to the eye, ear, or nose.
  • a suitable topical dose of active ingredient of a compound of the invention is 0.1 mg to 150 mg administered one to four, preferably one or two times daily.
  • the active ingredient may comprise from 0.001 % to 10% w/w, e.g. from 1 % to 2% by weight of the formulation, preferably not more than 5% w/w, and more preferably from 0.1 % to 1 % of the formulation.
  • the method comprises administering to the subject an initial daily dose of about 0.1 to 500 mg of a compound of formula (A) and increasing the dose by increments until clinical efficacy is achieved. Increments of about 5, 10, 25, 50, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.
  • the compounds of formula (A) may be prepared using the methods disclosed herein and routine modifications thereof, which will be apparent given the disclosure herein and methods are well known in the art. Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of representative compounds described herein may be accomplished as described in the following examples. If available, reagents may be purchased commercially, e.g., from Sigma Aldrich or other chemical suppliers.
  • solvent refers to a solvent inert under the conditions of the reaction being described in conjunction therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran (“THF”), dimethylformamide (“DMF”), chloroform, dichloromethane (DCM), diethyl ether, methanol, pyridine and the like.
  • solvents used in the reactions of the present invention are inert organic solvents, and the reactions are carried out under an inert gas, preferably nitrogen.
  • This Example compares the biological activity and hepatocyte stability of the compounds of formula (A) to4-amino-2-hydrogen pyrimidine analogs D-F such as compounds having the following structure (Compounds D-F were reported in patent US2013/0267524 and compound D is a close analog for reference purpose).
  • Enzymatic activity of different PI3K isoforms was measured to compare the PI3K isoform selectivity of the tested compounds, particularly selectivity of PI3K delta. Hepatocyte stability was also measured to assess the potential half-life of the tested compounds in human subjects.
  • Enzymatic activity of the class I PI3K isoforms in the presence of the compounds of Table 1 above was measured using a time-resolved fluorescence resonance energy transfer (TR- FRET) assay.
  • TR- FRET time-resolved fluorescence resonance energy transfer
  • the TR-FRET assay can monitor formation of the product 3,4,5-inositol triphosphate molecule (PIP3) as it competed with f uorescently labeled PIP3 for binding to the GRP-1 pleckstrin homology domain protein.
  • PIP3 3,4,5-inositol triphosphate molecule
  • An increase in phosphatidylinositide 3-phosphate product results in a decrease in TR-FRET signal as the labeled f uorophore is displaced from the GRP-1 protein binding site.
  • the PI3K isoforms were assayed under initial rate conditions in the presence of 10 ⁇ ATP, and compounds were tested in 10-dose IC50 mode starting at a concentration of 0.5 ⁇ . Control compound, PI- 103, was tested in 10-dose IC50 with 3-fold serial dilution starting at 10 ⁇ .
  • the hepatocyte assay was used to evaluate the metabolic stability of test articles (TA) following incubation in cryopreserved hepatocytes by monitoring parent drug disappearance via LC/MC.
  • the TA with 1% final DMSO concentration was incubated with 0.5 million hepatocytes/ml at 1 ⁇ substrate in duplicate. The incubation was carried out at 37°C with 5% C0 2 and saturating humidity. Samples were taken at 0, 1, 2, and 3 hours to monitor the disappearance of TA and a half-life (t 1/2) was determined. Table 2 below summarizes the tl/2 values (e.g. 1 1/2) collected from this Example.
  • Compound 2 was dosed by both iv and PO in the rats. Shown in Table 4 is a comparison of PK profiles of 2 and marketed PI3K delta inhibitor idelalisib, also known as 101 (PK data from idelalisib NDA filing): Compound 2 showed remarkably increased oral exposure after oral dose (10 mg/kg) in the SD rats. Oral exposure corrected with dose
  • AUCinf/dose is 20 fold higher than that of idelalisib.
  • Day 0 cell seeding for all cell lines with the density of 10000 cells /80 ⁇ ⁇ /well.
  • the final DMSO concentration of each well treated with test compound and corresponding vehicle control is 0.1%. Incubate the plate for 72 h in specified incubator.

Abstract

The present disclosure provides selective phosphoinositide 3-kinase inhibitors of formula (A), or pharmaceutically acceptable salts thereof. These compounds are useful for the treatment of conditions mediated by one or more P13K isoforms, such as PI3K delta (PI3K?). The present disclosure further provides methods of inhibiting phosphoinositide 3-kinase inhibitors using these compounds for treatment of disorders related to phosphatidylinositol 3-kinase activity.

Description

QUINOLINE ANALOGS AS PHOSPHATIDYLINOSITOL 3-KINASE INHIBITORS
CROSS-REFERENCE TO RELATED APPLICATIONS
[001] This application claims the benefit of U.S. Provisional Patent Application Serial No. 6/2304148, filed on March 4, 2016, the disclosure of which is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
[002] The present disclosure relates generally to quinoline analogs as inhibitors of
phosphatidylinositol 3-kinase (PI3K) activity. More specifically, the invention further relates to the preparation of the disclosed PI3K inhibitor analogs and their use in pharmaceutical compositions for the treatment of various diseases, conditions and disorders related to PI3K activity.
BACKGROUND OF THE INVENTION
[003] The class I phosphoinositide 3-kinases (PI3Ks) regulate phosphatidylinositol 4,5- bisphosphate (PIP2) phosphorylation. PI3K Converts PIP2 to the scaffolding binding element phosphatidylinositol (3,4,5)-trisphosphate (PIP3). PIP3 plays a key regulatory role in cell survival, signal transduction, control of membrane trafficking and other functions. (Di Paolo, G. et al. Nature 2006, 443, 651; Parker, P. J. et al. Biochem. Soc. Trans. 2004, 32, 893; Hawkins, P. T. et al. Biochem. Soc. Trans. 2006, 34, 647; Schaeffer, E. M. et al. Curr. Opin. Immnunol. 2000, 12, 282). Its dysregulation leads to various disease states such as cancer, inflammatory and autoimmune disorders.
[004] The Class I PI3Ks consist of four kinases further delineated into 2 subclasses. Class 1A PI3Ks consist of three closely related kinases, PBKoc, β, and δ existing as heterodimers composed of a catalytic subunit (pi 10a, β or δ) and one of several regulatory subunits. They generally respond to signaling through receptor tyrosine kinases (RTKs). ΡΙ3Κγ single class IB isoform, responds mainly to G-protein coupled receptors (GPCRs), and is composed of a pi 10γ catalytic subunit and one of two distinct regulatory subunits. PBKoc and ΡΒΚβ are ubiquitously expressed throughout a wide variety of tissue and organ types. PDKy is found mainly in leukocytes, but also in skeletal muscle, liver, pancreas, and heart (Cantly, C. Science 2002, 1655). The expression pattern of PI3K5 is restricted, to spleen, thymus, and peripheral blood leukocytes (Knight, Z. et al. Cell 2006, 125, 733).
[005] PI3K5 has been implicated as a major player in the adaptive immune system due to its expression pattern and evidence accumulated with genetically modified mice. Recently, activated PI3K delta syndrome (APDS) was described, a primary immunodeficiencies (PID) associated with a dominant gain-of-function mutation in which lysine replaced glutamic acid at residue 1021 (E1021K) in the pi 10δ protein. APDS was characterized by recurrent respiratory infections, progressive airway damage, and lymphopenia (Ivan Angulo et al. Science 2013, 342, 866). ΡΒΚδ inhibitors can potentially be supplemental to the treatments of B-cell related diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) by the biologies rituximab (Rituxan) and belimumab (Benlysta), as well as primary immunodeficiencies (PID). Several PI3K5 selective inhibitors, such as, idelalisib (GS-1101), IPI-145 and AMG 319 have entered the clinic targeting hematological malignancies, but few inhibitors have entered clinical trials for anti-inflammatory treatments (Cushing, T. et al. J Med Chem. 2015, 58, 480).
[006] In July 2014, the FDA and EMA granted first-in class PI3K delta inhibitor idelalisib approval to treat different types of leukemia; its safety and effectiveness to treat relapsed FL and relapsed SLL were established in a clinical trial with patients with indolent non-Hodgkin lymphomas. ("FDA approves Zydelig for three types of blood cancers". Food and Drug
Administration. July 23, 2014). However, the U.S. label for idelalisib has a boxed warning describing toxicities that can be serious and fatal, including liver toxicity. Fatal and/or serious hepatotoxicity occurred in 18% of patients treated with idelalisib monotherapy and 11% of patients treated with idelalisib in combination trials. Elevations in ALT or AST are greater than 5 times the upper limit of what has occurred normally. The liver toxicity may be related to the inhibition and induction of CYP enzymes by idelalisib and its metabolite GS-563117.
O¾ttp://www.accessdata.fda.gov/d.mgsatfda docs/nda/201.4/206545Origls000ClinPh.armR.pd.f).
[007] More recently, it was reported that pi 105 inactivation in mice protects against a broad range of cancers, including non-haematological solid tumors, and that pi 105 inactivation in regulatory T cells unleashes CD8(+) cytotoxic T cells and induces tumor regression. Thus, pi 105 inhibitors can break tumor-induced immune tolerance and potentially have wide usage in oncology. (Ali, et al., Nature: 2014, 510, 407-411). There still remains an unmet need for optimal PI3K delta inhibitors. For example, PI3K delta inhibitor can have improved in vivo stability to overcome the liability of inhibition or induction of CYP enzymes; an ideal PI3K delta inhibitor can have the potential of combination treatment of malignant tumors with other anti-cancer interventions, such as emerging immunotherapies. The present invention provides novel compounds that are inhibitors of PI3K isoforms with significantly improved profiles.
SUMMARY
[008] Compounds and pharmaceutically acceptable salts, prodrug, or solvate thereof useful for inhibiting PI3K isoforms, such as PI3K delta, are described herein. Compositions, including pharmaceutical compositions that include the compounds are also provided, as are methods of using and making the compounds. The compounds provided herein may find use in treating diseases, disorders, or conditions that are mediated by PI3K isoforms, such as PI3K delta.
[009] In one aspect, provided is a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate wherein:
Figure imgf000004_0001
Formula (A)
[010] X is N or CH;
[011] Each Ri and R2 is independently H, F, or S02Me.
[012] R3 is F or CI. [013] In one embodiment of formula (A) where X is N, both Ri and R2 are H, R3 is 7-F. The compound is of compound (2) in Table 1; or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
Figure imgf000005_0001
(2)
[014] In another embodiment of formula (A) where X is C, Ri is H, R2 is 2-S02Me, R3 is 8-F, the compound is compound (7) in Table 1, or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
Figure imgf000005_0002
[015] Also provided is a method of treating PI3K-mediated conditions or disorders with a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof. Further provided is a method of treatments of inflammatory diseases, such as asthma, chronic obstructive pulmonary disease, rheumatoid arthritis, multiple sclerosis, and lupus.
[016] Also provided is a method of inhibiting the growth or proliferation of cancer cells comprising contacting the cancer cells with an effective amount of a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate. Also provided is a method for increasing sensitivity of cancer cells to chemotherapy, comprising administering to a patient undergoing chemotherapy with a chemo therapeutic agent and a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate, sufficient to increase the sensitivity of cancer cells to the chemotherapeutic agent.
[017] Also provided are articles of manufacture that include a compound of formula (A), or a pharmaceutically acceptable, or prodrug, or solvate thereof.
DETAILED DESCRIPTION
[018] It is intended and understood that each and every variation of Ri and R2 may be combined with each and every variation of X as described for formula (A), as if each and every combination is individually described.
PI3K Inhibitor Compounds
[019] Provided herein are compounds that function as PI3K inhibitors. In one aspect, provided is a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate wherein:
Figure imgf000006_0001
Formula (A)
[020] X is N or CH;
[021] Each Ri and R2 are independently H, F, or S02Me.
[022] R3 is F or CI. [023] In one embodiment of formula (A) where X is N, both Ri and R2 are H, R3 is 7-F. The compound is of compound (2) in Table 1; or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
Figure imgf000007_0001
[024] In another embodiment of formula (A) where X is C, Ri is H, R2 is 2-S02Me, R3 is 8-F, the compound is compound (7) in Table 1, or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
Figure imgf000007_0002
Table 1. Re resentative Quinoline Compounds of Formula (A)
Figure imgf000008_0001
[025] Provided are also compounds of formula (A), or pharmaceutically acceptable salts, prodrugs, or solvates thereof. In certain embodiments, provided herein are also crystalline and amorphous forms of the compounds of formula (A), or pharmaceutically acceptable salt, prodrugs, or solvents thereof. [026] "Pharmaceutically-acceptable salt" means a salt prepared by conventional means, and are well known by those skilled in the art. The "pharmacologically acceptable salts" include basic salts of inorganic and organic acids (Berge et al., J. Pharm. Sci. 1977, 66: 1).
[027] A "solvate" is formed by treating a compound in a solvent. Solvates of salts of the compounds of formula (A) are also provided. In the case of treating compounds with water, the solvate is hydrates. Hydrates of the compounds of formula (A) are also provided.
[028] A "prodrug" includes any compound that converts into a compound of formula (A), when administered to a subject, e.g., upon metabolic processing of the prodrug.
Therapeutic Uses of the Compounds
[029] The compounds of formula (A), or pharmaceutically acceptable salt, prodrug, or solvate thereof may be used for treating PI3K mediated diseases or disorders. In one embodiment, provided are methods for inhibiting PI3K delta activity using a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof. In another embodiment, PI3K delta and gamma isomers may both be inhibited to achieve optimal efficacy.
[030] In addition to the therapeutic uses described herein, selected compounds of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof, have improved properties in at least one of the following parameters: (i) human hepatocyte stability, and (ii) pharmacokinetic profiles (PK) including oral exposure.
[031] In another embodiment, selected compounds of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof have improved human hepatocyte stability. Human hepatocyte stability in many cases correlates with pharmacokinetics in human better than the corresponding rodent pharmacokinetic studies. According to some embodiments, selected compounds may have hepatocyte stability of a half-life of greater than 24 hours.
[032] As used herein, "treat" or "treating" in reference to a disorder means to ameliorate or prevent the disorder or one or more of the biological manifestations of the disorder, to interfere with one or more points in the biological cascade that leads to or is responsible for the disorder, to alleviate one or more of the symptoms or effects associated with the disorder. As indicated above, "treatment" of a disorder includes prevention of the disorder, and "prevention" is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a disorder or biological manifestation thereof, or to delay the onset of such disorder or biological manifestation thereof. [033] "Subject" refers to a human (including adults and children) or other animal. In one embodiment, "patient" refers to a human.
[034] As used herein, "safe and effective dose" in reference to a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof an amount sufficient to treat the patient's condition but low enough to avoid serious side effects. A safe and effective dose of a compound will vary with the particular compound chosen (e.g. consider the potency, efficacy, and half-life of the compound); the route of administration chosen; the disorder being treated; the severity of the disorder being treated; the age, size, weight, and physical condition of the patient being treated; the medical history of the patient to be treated; the duration of the treatment; the nature of concurrent therapy; the desired therapeutic effect; and like factors.
[035] "Inhibition of PI3K delta activity" or variants refer to a decrease in PI3K delta activity as a direct or indirect response to the presence of a compound of formula (A), or a
pharmaceutically acceptable salt, prodrug, or solvate thereof, relative to the activity of PI3K delta in the absence of the compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
[036] The term "PI3K delta selective inhibitor" generally refers to a compound that inhibits the activity of the PI3K delta isoform more effectively than other isoforms of the PI3K family (e.g., PI3K alpha, beta, or gamma).
[037] The potencies of compounds as inhibitors of an enzyme activity (or other biological activity) can be established by determining the concentrations at which each compound inhibits the activity to a predefined extent and then comparing the results. "IC50" or "IC90" of an inhibitor can be determined by the concentration that inhibits 50% or 90% of the activity in a biochemical assay, which can be accomplished using conventional techniques known in the art, including the techniques describes in the Examples below.
[038] PI3K delta is expressed primarily in hematopoietic cells including leukocytes such as T- cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages. Due to its integral role in immune system function, PI3K delta is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases. By inhibiting aberrant proliferation of hematopoietic cells, PI3K delta inhibitors can ameliorate the symptoms and secondary conditions that result from a primary effect such as excessive system or localized levels of leukocytes or lymphocytes.
[039] In one aspect, the invention thus provides a method of treating a disorder mediated by inappropriate P13-kinase activity comprising administering a safe and effective dose of a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate thereof [040] In one embodiment, PI3K mediated diseases or disorders are selected from the group consisting of respiratory diseases (including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF)); allergic diseases (including allergic rhinitis and atopic dermatitis); autoimmune diseases (including rheumatoid arthritis and multiple sclerosis); inflammatory disorders (including inflammatory bowel disease); hematologic malignancies; solid tumors; neurodegenerative diseases; pancreatitis; kidney diseases;
transplantation rejection; graft rejection; lung injuries
[041] In one embodiment, the compounds described herein may be used to treat cancers that are mediated by inappropriate PI3K delta activity. In certain embodiments, the disease is a hematologic malignancy. In certain embodiments, the disease is lymphoma, such as Burkitt lymphoma, diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL), follicular lymphoma, lymphoplasmacytic lymphoma, Waldenstrom macroglobulinemia, and marginal zone lymphoma. In one embodiment, the disorder is multiple myeloma, or leukemia, such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic
lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disease (MPD), chronic myeloid leukemia (CML).
[042] In other embodiments, the disease is a solid tumor. In particular embodiments, the indication is to treat solid tumor with abnormal PI3K delta expression, such as pancreatic cancer, gastric cancer, esophageal cancer, and breast cancer. In some embodiment, the compounds alone or with combination of other anti-cancer therapies may be used to treat prostate cancer, bladder cancer, colorectal cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, head and neck cancer, melanoma, neuroendocrine cancers, brain tumors, bone cancer, or soft tissue sarcoma.
[043] In some embodiments, PI3K mediated diseases or disorders are severe autoimmune disease as asthma, type I diabetes, rheumatoid arthritis, multiple sclerosis, chronic obstructive pulmonary disease (COPD), and lupus. Combination Therapies
[044] In one embodiment, a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat cancers or inflammatory disorders. The one or more additional therapeutic agents may be a chemotherapeutic agent, a radiotherapy, a targeted therapy, an immunotherapeutic agent or any current best of care treatment, either as a small molecule or a biologic nature.
[045] Targeted therapies include but not limit to an inhibitor to cyclin-dependent kinase (CDK) such as CDK1, CDK2, CDK4/6, CDK7, and CDK9, Janus kinase (JAK) such as JAK1, JAK2 and/or JAK3 , spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), mitogen- activated protein kinase (MEK) such as MEK 1 and MEK2, bromodomain containing protein inhibitor (BRD) such as BRD4, isocitrate dehydrogenase (IDH) such as IDH1, histone deacetylase (HDAC), or any combination thereof.
[046] Chemotherapeutic agents may be categorized by their mechanism of action into:
alkylating agents, antimetabolites, anti-microtubule agents, topoisomerase inhibitors and cytotoxic agents. A compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with chemotherapeutic s to sensitize and improve the efficacy of certain chemotherapeutic agents to treat blood or solid tumors.
[047] The immunotherapeutic agents include and are not limited to therapeutic antibodies, small molecules and vaccines suitable for treating patients; such as IDOl and TD02 inhibitors, A2A receptor inhibitors, arginase inhibitors, toll-like receptor agonists, chemokine regulators (including CCR and CXCR families), check point blockage antibodies such as antibodies that regulate PD-1, PD-Ll, CTLA-4, OX40-OX40 ligand, LAG3, TIM3, or any combination thereof.
[048] Radiotherapy is part of cancer treatment to control or kill malignant cells and commonly applied to the cancerous tumor because of its ability to control cell growth. A compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with radiotherapy, to improve the efficacy of radiotherapy to treat blood or solid tumors, or with surgery, chemotherapy, immunotherapy and combination of the four.
[049] In certain embodiments, a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat patients who are substantially refractory to at least one chemotherapy treatment, or in relapse after treatment with chemotherapy.
Pharmaceutical Compositions
[050] In another aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate salt thereof and a pharmaceutically acceptable carrier or excipient. The pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and topical administration, etc.
[051] Compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and can be prepared in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules. Oral composition may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. The tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
[052] Certain injectable compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
[053] Suitable compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier. Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and
predetermined rate over a prolonged period of time, and means to secure the device to the skin. [054] Suitable compositions for topical application, e.g., to the skin and eyes, include aqueous solutions, suspensions, ointments, creams, gels or spray formulations, e. g., for delivery by aerosol or the like. Such topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like.
[055] As used herein a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray, atomizer or nebulizer, with or without the use of a suitable propellant.
[056] The invention further provides pharmaceutical compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose. Such agents, which are referred to herein as "stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
Modes of Administration and Dosing
[057] The pharmaceutical compositions may be administered in either single or multiple doses. A compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate salt thereof can be formulated so as to provide the desired release schedule of the active ingredient based on the therapeutic treatment purpose.
[058] The pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules. For example, these may contain an amount of active ingredient from about 0.1 to 1000 mg, preferably from about 0.1 to 500 mg. A suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods. The daily dose can be administered in one to four doses per day. For therapeutic purposes, the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration drops suitable for administration to the eye, ear, or nose. A suitable topical dose of active ingredient of a compound of the invention is 0.1 mg to 150 mg administered one to four, preferably one or two times daily. For topical administration, the active ingredient may comprise from 0.001 % to 10% w/w, e.g. from 1 % to 2% by weight of the formulation, preferably not more than 5% w/w, and more preferably from 0.1 % to 1 % of the formulation.
[059] In a particular embodiment, the method comprises administering to the subject an initial daily dose of about 0.1 to 500 mg of a compound of formula (A) and increasing the dose by increments until clinical efficacy is achieved. Increments of about 5, 10, 25, 50, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.
Synthesis of the Compounds of Formula (A)
[060] The compounds of formula (A) may be prepared using the methods disclosed herein and routine modifications thereof, which will be apparent given the disclosure herein and methods are well known in the art. Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of representative compounds described herein may be accomplished as described in the following examples. If available, reagents may be purchased commercially, e.g., from Sigma Aldrich or other chemical suppliers.
General
[061] Reagents and solvents used below can be obtained from commercial sources. 1H-NMR spectra were recorded on a Mercury 300 MHZ NMR spectrometer. Significant peaks are tabulated in the order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br s, broad singlet), coupling constant(s) in Hertz (HZ) and number of protons. Mass spectrometry results are reported as the ratio of mass over charge, followed by the relative abundance of each ion (in parentheses Electrospray ionization (ESI) mass spectrometry analysis was conducted on an Agilent 1100 series LC/MSD electrospray mass spectrometer. All compounds could be analyzed in the positive ESI mode using acetonitrile water with 0.1% TFA as the delivery solvent.
Synthetic Reaction
[062] The terms "solvent", "inert organic solvent", or "inert solvent" refer to a solvent inert under the conditions of the reaction being described in conjunction therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran ("THF"), dimethylformamide ("DMF"), chloroform, dichloromethane (DCM), diethyl ether, methanol, pyridine and the like. Unless specified to the contrary, the solvents used in the reactions of the present invention are inert organic solvents, and the reactions are carried out under an inert gas, preferably nitrogen.
[063] Preparation of 8-substituted quinoline amines:
Figure imgf000016_0001
Example 1: (S)-l-(8-Fluoro-2-pyridin-2-yl-quinolin-3-yl)-ethylamine Step 1:
Figure imgf000016_0002
[066] To a solution of 2-chloro-8-fluoroquinoline-3-carboxaldehyde (1.5 g, 7.2 mmol) in anhydrous THF (20 mL) was added titanium isopropoxide (4.3 mL, 1.4 mmol) at r.t. After 15 minutes, (R)-2-methyl-2-propanesulfinamide (0.867 g, 7.2 mmol) was added and stirring was continued overnight at r.t. Water (100 mL) was added to the reaction mixture and the precipitate obtained was filtered and washed with DCM. The organic layer was dried (Na2S04), filtered and concentrated in vacuo to give the crude material as a pale yellow solid which was purified by column chromatography on silica gel (EtOAc/hexane, 4/5)) to give a pale yellow solid (2.0 g, 89%). Mass Spectrum (ESI) m/e: 313 (M+l).
[067] Step 2:
Figure imgf000017_0001
[068] To a solution of 2-Methyl-propane-2-sulfinic acid 2-chloro-8-fluoro-quinolin-3- ylmethyleneamide (0.95 g, 2.8 mmol) in DCM (22 mL) was added dropwise MeMgCl (1.94 mL, 5.8 mmol; 3 M in THF) over 10 minutes at -78 °C under nitrogen. The reaction mixture was allowed to reach r.t. with stirring overnight. The mixture was cooled in ice-salt as saturated aqueous NH4C1 (50 mL) was slowly added with stirring. The aqueous layer was extracted with DCM (2 x 50 mL). The organic layer was dried (MgS04) filtered and concentrated in vacuo to give a yellow oil which was purified by column chromatography on silica gel (EtOAc/hexane, 4/5 to EtOAc) to give a pale yellow solid (260 mg, 28%). Mass Spectrum (ESI) m/e: 329 (M+1).
[069] Step 3:
Figure imgf000017_0002
[070] A mixture of (S)-2-Methyl-propane-2-sulfinic acid [l-(2-chloro-8-fluoro-quinolin-3- yl)-ethyl] -amide (0.186 g, 0.57 mmol), Pd(PPh3)4 (0.066 g, 0.057 mmol, 0.1 eq) and 2- (tributylstannyl)-pyridine (0.51 g, 1.4 mmol, 2.4 eq) in dioxane (5 mL) was heated to 110°C under N2. After stirring overnight, the combined solvents were concentrated and purified by column chromatography on silica gel (EtOAc) to give 2-Methyl-propane-2- sulfuric acid [l-(8- fluoro-2-pyridin-2-yl-quinolin-3-yl)-ethyl]-amide (160 mg, 43%). Mass Spectrum (ESI) m e: 372 (M+1).
[071] To a solution of the above material (160 mg) in MeOH (2 mL) was added 4 N HC1 in dioxane (2 mL) at rt and the resulted reaction mixture was stirred for 2 hours and concentrated under reduced pressure. Ethyl ether was added and sonicated for 2 min and filtered to give (S)-l- (8-fluoro-2-pyridin-2-yl-quinolin-3-yl)-ethylamine as HC1 salt. Mass Spectrum (ESI) m/e: 268 (M+l).
[072] (S)- l-(8-Chloro-2-pyridin-2-yl-quinolin-3-yl)-ethylamine (10) and (S)-l-[8-Fluoro-2- (2-methanesulfonyl-phenyl)-quinolin-3-yl]-ethylamine (11) were prepared in the similar manner.
[073] Preparation of 7-substituted quinoline amines:
Figure imgf000018_0001
[074] Example 2: (S)-l-(7-Fluoro-2-phenyl-quinolin-3-yl)-ethylamine (12)
Figure imgf000018_0002
[075] Compound (S)-2-(l-(2-Chloro-7-fluoroquinolin-3 -yl)ethyl)isoindoline-l,3-dione was prepared according to the literature (J. Med. Chem. 2015, 58, 480-511). This compound (280 mg, 0.79 mmol), phenylboronic acid (146 mg, 1.2 mmol), and potassium carbonate (328 mg, 2.4 mmol) were combined in 6 mL of anhydrous DMF under an atmosphere of N2. The solution was purged with N2 for ~5 min before adding PdC12(dppf)DCM (64 mg, 0.079 mmol). The solution was heated at 100° C for 3 h, and then cooled to 50° C. The solution was concentrated under vacuum to give a brownish residue, which was diluted with EtOAc (12 mL). The organic layers were then washed with water (3x3 mL), followed by brine (10 mL). The combined aq. layers were extracted with DCM (3x2 mL). The combined organic layers were dried over MgS04 and then concentrated under vacuum. The residue obtained was purified by silica gel flash chromatography eluting with a gradient of 20% to 40% EtOAc/hexane. The fractions containing the pure product were combined and concentrated under vacuum to give (S)-2-[l-(7-fluoro-2- phenyl-quinolin-3-yl)-ethyl]-isoindole-l,3-dione (263 mg, 84% yield) as a light yellow foam. Mass Spectrum (ESI) m/e: 397 (M+l).
[076] To a slurried suspension of (S)-2-[l-(7-fluoro-2-phenyl-quinolin-3-yl)-ethyl]-isoindole- 1,3-dione (260 mg, 0.65 mmol) in anhydrous ethanol (3 mL) was added NH2NH2 (0.11 g, 5.0 eq) dropwise. The reaction mixture was heated to 90° C for 30 min and cooled to rt. The reaction mixture was filtered and washed with EtOAc. The resulting EtOAc solution was washed with water, brine and dried over Na2S04. Removal of solvents gave a tan oil of (S)-l-(7-fluoro-2- phenyl-quinolin-3-yl)-ethylamine (122 mg, 71%). Mass Spectrum (ESI) m/e: 267 (M+l).
[077] (S)-l-[2-(3,5-Difluoro-phenyl)-7-fluoro-quinolin-3-yl]-ethylamine (14), (S)-l-[7- Fluoro-2-(2-methanesulfonyl-phenyl)-quinolin-3-yl]-ethylamine (15), and (S)-l-[7-Fluoro-2-(3- fluoro-phenyl)-quinolin-3-yl]-ethylamine (16) were prepared in the similar manner.
[078] Exampe 3: (S)-l-(7-fluoro-2-(pyridin-2-yl) quinolin-3-yl)ethanamine (13)
Figure imgf000019_0001
[079] A mixture of (S)-2-(l-(2-chloro-7-fluoroquinolin-3-yl)ethyl)isoindoline-l,3-dione (0.86 g, 2.4 mmol), Pd(PPh3)4 (0.28 g, 0.24 mmol, 0.1 eq) and 2-(tributylstannyl)-pyridine (1.07 g, 2.9 mmol, 1.2 eq) in dioxane (30 mL) was heated to 90°C under N2. After stirring overnight, LC- MS showed 30% completion. The reaction mixture was heated to 101° C for additional 2 days. The reaction mixture was then cooled to rt and the resulted solid was filtered and washed with EtOAc to give a tan solid of 2-((S)-l-(7-fluoro-2(pyridin-2-yl)quinolin-3-yl)ethyl)isoindoline- 1,3-dione was obtained (0.84 g, 88%). Mass Spectrum (ESI) m/e: 398 (M+l). [080] To a slurried suspension of 2-((S)-l-(7-fluoro-2-(pyridin-2-yl)quinolin-3- yl)ethyl)isoindoline-l,3-dione (0.84 g, 2.1 mmol) in anhydrous ethanol (5 mL) was added NH2NH2 (0.34 g, 10.4 mmol) dropwise. The reaction mixture was heated to 90° C for 30 min and cooled to rt. The reaction mixture was filtered and washed with EtOAc. The resulting EtOAc solution was washed with water, brine and dried over Na2S04. Removal of solvents gave a tan oil of (S)-l-(7-fluoro-2-(pyridin-2-yl)quinolin-3-yl)ethanamine (399 mg, 71%). Mass Spectrum (ESI) m/e: 268 (M+l).
[081] Preparation of 2,4,6-Triamino-pyrimidine-5-carbonitrile
Figure imgf000020_0001
[082] Ammonium hydroxide (4 mL) was added to a solution of 2,4,6-trichloropyrimidine-5- carbonitrile (1.0 g, 4.8 mmol) in dioxane (4 mL) at room temperature. The solution was warmed to 50°C and stirred for 3 hrs. The reaction mixture was cooled to 10°C and water (10 mL) was added. The resulting solid was filtered, washed with water, and dried under high vacuum to afford 2,4,6-triamino-pyrimidine-5 carbonitrile as a white solid (0.8 g). Mass Spectrum (ESI) m/e: 170 (M+H).
[083] Preparation of (S)-2,4-diamino-6-{ l-[7-fluoro-2-(3-fluoro-phenyl)-quinolin-3-yl]- ethylamino } -pyrimidine-5-carbonitrile (5)
Figure imgf000020_0002
[084] Potassium fluoride (34 mg, 0.58 mmol) was added to a solution of (S)-l-[7-fluoro-2-(3- fluoro-phenyl)-quinolin-3-yl]-ethylamine (90 mg, 0.31 mmol) and 2,4-diamino-6- chloropyrimidine-5-carbonitrile (60 mg, 0.35 mmol) in diisopropylethylamine (0.1 mL, 0.60 mmol) and DMSO (2 mL). The resultant mixture was heated to 100°C for 14 hours, after which time the reaction was cooled to room temperature, and diluted with water (5 mL) and the resulted solid was filtered, washed with water and dried and purified by preparative TLC to give a white solid as (S)-2,4-diamino-6-{ l-[7-fluoro-2-(3-fluoro-phenyl)-quinolin-3-yl]-ethylamino}- pyrimidine-5-carbonitrile. Mass Spectrum (ESI) m/e: 418 (M+1). 1H NMR (300 MHz, DMSO- d6) δ ppm 8.49 (s, 1H), 8.10 (dd, J = 5.7, 3.0 Hz, 1H), 7.75 (dd, J = 11, 2.4 Hz, 1H), 7.52-7.60 (m, 3H), 7.27-7.32 (m, 1H), 7.12 (d, J = 7.2 Hz, 1H), 6.52 (s, 2H), 6.10 (s, br, 2H), 5.40-5.45 (m, 1H), 1.30 (d, J = 6.9 Hz, 3H).
[085] The following compounds were prepared in the similar manner.
[086] (S)-2,4-diamino-6-[l-(7-fluoro-2-phenyl-quinolin-3-yl)-ethylamino]-pyrimidine-5- carbonitrile (Compound 1, Table 1). Mass Spectrum (ESI) m/e: 400 (M+1). 1H NMR (300 MHz, DMSO-d6) 5 ppm 8.46 (s, 1H), 8.07 (dd, J = 6.6, 2.4 Hz, 1H), 7.67-7.70 (m, 2H), 7.49-7.52 (m, 3H), 7.13 (d, J = 7.5 Hz, 1H),6.52 (s, 2H), 6.10 (s, br, 2H), 5.45-5.50 (m, 1H), 1.27 (d, J = 6.9 Hz, 3H).
[087] (S)-2,4-Diamino-6-[l-(7-fluoro-2-pyridin-2-yl-quinolin-3-yl)-ethylamino]-pyrimidine- 5-carbonitrile (Compound 2, Table 1). Mass Spectrum (ESI) m/e: 401 (M+1). 1H NMR (300 MHz, DMSO-d6) 5 ppm 8.72 (s, 1H), 8.56 (m, 1H), 7.54-8.12 (m, 6H), 6.50 (s, 2H), 6.08 (s, br, 2H), 5.65-5.75 (m, 1H), 1.35 (d, J = 6.9 Hz, 3H).
[088] (S)-2,4-Diamino-6-{ l-[2-(3,5-difluoro-phenyl)-7-fluoro-quinolin-3-yl]-ethylamino}- pyrimidine-5-carbonitrile (Compound 3, Table 1). Mass Spectrum (ESI) m/e: 436 (M+1). 1H NMR (300 MHz, DMSO-d6) δ ppm 8.52 (s, 1H), 8.13 (dd, J = 5.7, 3.0 Hz, 1H), 7.76 (dd, J = 7.5, 1.8 Hz, 1H), 7.54-7.60 (m, 1H), 7.08-7.45 (m, 2H), 7.09 (d, J = 6.9 Hz, 1H), 6.52 (s, 2H), 6.12 (s, br, 2H), 5.35-5.40 (m, 1H), 1.34 (d, J = 6.3 Hz, 3H).
[089] (S)-2,4-Diamino-6-{ l-[7-fluoro-2-(2-methanesulfonyl-phenyl)-quinolin-3-yl]- ethylamino}-pyrimidine-5-carbonitrile (Compound 4, Table 1). Mass Spectrum (ESI) m/e: 478 (M+1). 1H NMR (300 MHz, DMSO-d6) δ ppm 8.50 (s, 1H), 8.13 (dd, J = 5.7, 3.0 Hz, 1H), 7.77- 7.89 (m, 4H), 7.56-7.61 (m, 1H), 7.06 (d, J = 7.5 Hz, 1H), 6.54 (s, 2H), 6.24 (s, br, 2H), 5.15- 5.25 (m, 1H), 3.37 (s, 3H), 1.27 (d, J = 6.9 Hz, 3H). [090] (S)-2,4-Diamino-6-[l-(8-fluoro-2-pyridin-2-y^
5-carbonitrile (Compound 6, Table 1). Mass Spectrum (ESI) m/e: 401 (M+l). 1H NMR (300 MHz, DMSO-d6) 5 ppm 8.51 (s, 1H), 8.16 (m, 1H), 7.54-8.02 (m, 6H), 6.51 (s, 2H), 6.01 (s, br, 2H), 5.23-5.34 (m, 1H), 1.30 (d, J = 6.9 Hz, 3H).
[091] (S)-2,4-Diamino-6-{ l-[8-fluoro-2-(2-methanesulfonyl-phenyl)-quinolin-3-yl]- ethylamino}-pyrimidine-5-carbonitrile (Compound 7, Table 1). Mass Spectrum (ESI) m/e: 478 (M+l). 1H NMR (300 MHz, MeOH-d4) 5 ppm 8.74 (d, J = 4.5 Hz, 1H), 8.51 (s, 1H), 8.03-7.94 (m, 2H), 7.80 (d, J = 8.4 Hz, 1H), 7.61-7.45 (m, 4H), 5.84 (q, J = 6.9 Hz, 1H), 4.60 (s, 1H), 3.33 (s, 3H), 1.41 (d, J = 6.9 Hz, 3H).
[092] (S)-2,4-Diamino-6-[l-(8-chloro-2-pyridin-2-yl-quinolin-3-yl)-ethylamino]-pyrimidine- 5-carbonitrile (Compound 8, Table 1). Mass Spectrum (ESI) m/e: 417 (M+l). 1H NMR (300 MHz, DMSO-d6) δ ppm 8.74 (d, J= 4.5 Hz, 1H), 8.60 (s, 1H), 8.06-7.94 (m, 3H), 7.75 (d, J = 8.4 Hz, 1H), 7.64-7.53 (m, 2H), 6.48 (s, 2H), 5.80-5.74 (m, 1H), 1.39 (d, J = 6.0 Hz, 3H).
Biological Examples
[093] Activity testing was conducted in the Examples below using methods described herein and those well known in the art.
Characterization of Compounds of Formula (A)
[094] This Example compares the biological activity and hepatocyte stability of the compounds of formula (A) to4-amino-2-hydrogen pyrimidine analogs D-F such as compounds having the following structure (Compounds D-F were reported in patent US2013/0267524 and compound D is a close analog for reference purpose).
Figure imgf000023_0001
[095] Enzymatic activity of different PI3K isoforms was measured to compare the PI3K isoform selectivity of the tested compounds, particularly selectivity of PI3K delta. Hepatocyte stability was also measured to assess the potential half-life of the tested compounds in human subjects.
[096] Each of these biological experiments are described below. [097] Enzymatic activity of PI3K isoforms
[098] Enzymatic activity of the class I PI3K isoforms in the presence of the compounds of Table 1 above was measured using a time-resolved fluorescence resonance energy transfer (TR- FRET) assay.
[099] The TR-FRET assay can monitor formation of the product 3,4,5-inositol triphosphate molecule (PIP3) as it competed with f uorescently labeled PIP3 for binding to the GRP-1 pleckstrin homology domain protein. An increase in phosphatidylinositide 3-phosphate product results in a decrease in TR-FRET signal as the labeled f uorophore is displaced from the GRP-1 protein binding site. [100] The PI3K isoforms were assayed under initial rate conditions in the presence of 10 μΜ ATP, and compounds were tested in 10-dose IC50 mode starting at a concentration of 0.5 μΜ. Control compound, PI- 103, was tested in 10-dose IC50 with 3-fold serial dilution starting at 10 μΜ.
[101] Data are normalized based on negative (DMSO) control. The alpha, beta, delta, and gamma IC50 values were calculated from the fit of the dose-response curves to a four parameter equation. IC50 are reported in units of nM.
[102] IC50 values were obtained for all PI3K isoforms (α, β, δ, and γ), and Table 2 summarizes the IC50 data collected for PI3K5 in this Example.
Hepatocyte stability
The hepatocyte assay was used to evaluate the metabolic stability of test articles (TA) following incubation in cryopreserved hepatocytes by monitoring parent drug disappearance via LC/MC. The TA with 1% final DMSO concentration was incubated with 0.5 million hepatocytes/ml at 1 μΜ substrate in duplicate. The incubation was carried out at 37°C with 5% C02 and saturating humidity. Samples were taken at 0, 1, 2, and 3 hours to monitor the disappearance of TA and a half-life (t 1/2) was determined. Table 2 below summarizes the tl/2 values (e.g. 1 1/2) collected from this Example.
Table 2.
Figure imgf000024_0001
[103] The results from this example demonstrate that compounds 1 and 4 of formula (A) have moderately improved stability in human hepatocytes (i.e. longer half-life), than compounds D and E (2.7x and 1.3x). However, several compounds showed surprisingly dramatic improvement for the stability in human hepatocytes (i.e. longer half-life); Table 3 below shows the comparison of t ½ of Compounds 2, 5, 7, H, G, and F. The dramatically improved stability of compounds in human hepatocytes will likely reduce the formation of drug metabolites in vivo and may contribute positively to the safety of the compounds in the clinical trials.
Table 3.
Figure imgf000025_0001
[104] Compound 2 was dosed by both iv and PO in the rats. Shown in Table 4 is a comparison of PK profiles of 2 and marketed PI3K delta inhibitor idelalisib, also known as 101 (PK data from idelalisib NDA filing): Compound 2 showed remarkably increased oral exposure after oral dose (10 mg/kg) in the SD rats. Oral exposure corrected with dose
(AUCinf/dose) is 20 fold higher than that of idelalisib.
Table 4
Figure imgf000026_0001
Comp 2 (po) 29491 2949 3173 31 7 2 3.12 NA \ NA 80
Cytotoxicity assay of compound 2 of Table 1 in selected cell lines [105] Procedure
[106] Day 0: cell seeding for all cell lines with the density of 10000 cells /80 μΐ^ /well.
[107] Day 1, drug treatment: Prepare 3-fold serial dilutions of compound stock solutions with growth medium. Dispense 20 μΐ^ (5x) d^g solution in each well (duplicates for each
concentration), the final DMSO concentration of each well treated with test compound and corresponding vehicle control is 0.1%. Incubate the plate for 72 h in specified incubator.
[108] Day 4, plate reading: Thaw CTG solution and equilibrate to room temperature, add 100 μL· of CTG per well, mix the contents for 2 min on the plate shaker, incubate for 10 min before recording the luminescence signal using Envision.
[109] Data analysis
[110] The Cell viability (percent of control= T/C, the optical density of the test well after a 3- day period of exposure to test drug is T, and the control optical density is C) of each dose were fitted using a nonlinear regression model with a sigmoidal dose response with GraphPad Prism version 5, and the IC50 that the concentration of test drug where 100 x T / C = 50 were calculated
Table 5.
ICso (nM)
Figure imgf000026_0002

Claims

CLIAMS WHAT IS CLAIMED IS :
1. A selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound having a structure of formula (A), or a pharmaceutically acceptable salt, or solvate thereof:
Figure imgf000027_0001
(A) wherein X is selected from the group consisting of N or CH;
each Ri and R2 is independently selected form the group consisting of H, F, and
S02Me, and
each R3 is independently selected from the group consisting of F and CI.
2. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 1, wherein R3 is independently selected for the group consisting of 7-F, 8-F and 8-Cl, or a pharmaceutically acceptable salt, or solvate thereof.
3. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 2, wherein X is N, each of Ri and R2 is H, or a pharmaceutically acceptable salt, or solvate thereof.
4. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 2, wherein X is CH and Ri is S02Me, or a pharmaceutically acceptable salt, or solvate thereof.
5. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 2, wherein X is CH and Ri is F, or a pharmaceutically acceptable salt, or solvate thereof.
6. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 2, wherein X is CH, Ri is H and R2 is H, or a pharmaceutically acceptable salt, or solvate thereof.
7. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 1, wherein X is N, each of Ri and R2 is H, and R3 is 7-F, or a pharmaceutically acceptable salt, or solvate thereof.
8. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 1, wherein X is CH, Ri is H, R2 is 2-S02Me, and R3 is 8-F, or a pharmaceutically acceptable salt, or solvate thereof.
9. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 1, wherein the compound's hepatocyte stability ranges from 6 to 59 Ti/2 (h), and wherein the compound has improved pharmacokinetics (PK) data than known PI3K5 inhibitors.
10. A method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase delta (PI3K5) comprising:
contacting kinase cells with an effective amount of a phosphoinositide 3-kinase delta (PI3K5) inhibitor compound having a structure of formula (A), or a pharmaceutically acceptable salt, or solvate thereof:
Figure imgf000028_0001
wherein X is selected from the group consisting of N or CH;
each Ri and R2 is independently selected form the group consisting of H, F, and
S02Me, and
each R3 is independently selected from the group consisting of F and CI.
11. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase delta (PI3K5) according to claim 10, wherein R3 is independently selected for the group consisting of 7-F, 8-F and 8-Cl, or a pharmaceutically acceptable salt, or solvate thereof.
12. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase delta (PI3K5) according to claim 10, wherein X is N, each of Ri and R2 is H, or a pharmaceutically acceptable salt, or solvate thereof.
13. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase delta (PI3K5) according to claim 10, wherein X is CH and Ri is S02Me, or a pharmaceutically acceptable salt, or solvate thereof.
14. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase delta (PI3K5) according to claim 10, wherein X is N, each of Ri and R2 is H, and R3 is 7-F, or a pharmaceutically acceptable salt, or solvate thereof.
15. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase delta (PI3K5) according to claim 10, wherein X is C, Ri is H, R2 is 2- S02Me, and R3 is 8-F, or a pharmaceutically acceptable salt, or solvate thereof.
16. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase delta (PI3K5) according to claim 10, wherein the compound has optimized hepatocyte stability varying from 6 to 59 T1/2 (h), and wherein the compound has improved pharmacokinetics (PK) data than known PI3K5 inhibitors.
17. A selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound having a structure of formula (A), or a pharmaceutically acceptable salt, or solvate thereof:
Figure imgf000030_0001
(A) wherein X is selected from the group consisting of N or CH;
Ri is H;
P2 is selected form the group consisting of H and 2-S02Me, and
P 3 is selected from the group consisting of 7-F and 8-F,
wherein the compound has optimized hepatocyte stability varying from 6 to 59
18. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 17, wherein X is N, R2 is H, and R3 is 7-F, or a pharmaceutically acceptable salt, or solvate thereof.
19. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 17, wherein X is CH, Rl is H, R2 is 2-S02Me, and R3 is 8-F, or a pharmaceutically acceptable salt, or solvate thereof.
20. The selective phosphoinositide 3-kinase delta (PI3K5) inhibitor compound of claim 17, wherein the compound has lowered cytotoxicity data than known PI3K5 inhibitors, and wherein the compound has improved pharmacokinetics (PK) data than known PI3K5 inhibitors.
PCT/US2017/019970 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors WO2017155741A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EP17763752.7A EP3400227B1 (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
US16/072,150 US10792283B2 (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
CA3013490A CA3013490C (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
KR1020187025588A KR102256497B1 (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
ES17763752T ES2865424T3 (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
KR1020217014690A KR20210059033A (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
CN201780004233.0A CN108290898B (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
JP2018533215A JP7201992B2 (en) 2016-03-05 2017-02-28 Quinoline analogues as phosphatidylinositol 3-kinase inhibitors
US17/004,734 US11446301B2 (en) 2016-03-05 2020-08-27 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662304148P 2016-03-05 2016-03-05
US62/304,148 2016-03-05

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/072,150 A-371-Of-International US10792283B2 (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
US17/004,734 Continuation US11446301B2 (en) 2016-03-05 2020-08-27 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Publications (1)

Publication Number Publication Date
WO2017155741A1 true WO2017155741A1 (en) 2017-09-14

Family

ID=59789651

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/019970 WO2017155741A1 (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Country Status (8)

Country Link
US (2) US10792283B2 (en)
EP (1) EP3400227B1 (en)
JP (1) JP7201992B2 (en)
KR (2) KR20210059033A (en)
CN (2) CN108290898B (en)
CA (2) CA3124815A1 (en)
ES (1) ES2865424T3 (en)
WO (1) WO2017155741A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10792283B2 (en) 2016-03-05 2020-10-06 Hangzhou Zhengxiang Pharmaceuticals Co., Ltd. Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022188758A1 (en) * 2021-03-09 2022-09-15 南京征祥医药有限公司 Quinoline compound salt or crystal form, preparation method therefor, and application thereof
CN114276326B (en) * 2021-03-09 2022-11-11 南京征祥医药有限公司 Salt of quinoline compound, preparation method and application
CN114276329B (en) * 2021-03-09 2022-11-11 南京征祥医药有限公司 Crystal form of salt of quinoline compound, preparation method and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100331306A1 (en) * 2009-06-25 2010-12-30 Amgen Inc. Heterocyclic compounds and their uses
WO2012037204A1 (en) * 2010-09-14 2012-03-22 Exelixis, Inc. Inhibitors of pi3k-delta and methods of their use and manufacture
WO2012061696A1 (en) * 2010-11-04 2012-05-10 Amgen Inc. 5 -cyano-4, 6 -diaminopyrimidine or 6 -aminopurine derivatives as pi3k- delta inhibitors
US20130267524A1 (en) 2012-04-04 2013-10-10 Amgen Inc. Heterocyclic compounds and their uses
US20160016915A1 (en) * 2012-12-21 2016-01-21 Gilead Calistoga Llc Phosphatidylinositol 3-kinase inhibitors

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2680853C (en) * 2007-03-23 2012-07-17 Amgen Inc. 3- substituted quinoline or quinoxaline derivatives and their use as phosphatidylinositol 3-kinase (pi3k) inhibitors
EP2137186B1 (en) * 2007-03-23 2016-01-27 Amgen Inc. Heterocyclic compounds and their uses
US20110135655A1 (en) 2009-01-13 2011-06-09 PHILADELPHIA HEALTH AND EDUCATION CORPORATION d/b/a Drexel University College of Medicine; Role of PI3K p110 delta Signaling in Retroviral Infection and Replication
TW201414734A (en) * 2012-07-10 2014-04-16 Takeda Pharmaceutical Azaindole derivatives
TR201806822T4 (en) 2013-06-14 2018-06-21 Gilead Calistoga Llc Phosphatidylinositol 3-kinase inhibitors.
AU2014354769A1 (en) 2013-11-26 2016-05-26 Gilead Sciences, Inc. Therapies for treating myeloproliferative disorders
WO2015106014A1 (en) * 2014-01-09 2015-07-16 Takeda Pharmaceutical Company Limited Azaindole derivatives
US9637488B2 (en) * 2015-01-29 2017-05-02 Fuqiang Ruan Heterocyclic compounds as inhibitors of class I PI3KS
CA3124815A1 (en) 2016-03-05 2017-09-14 Hangzhou Zhengxiang Pharmaceuticals Co., Ltd. Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100331306A1 (en) * 2009-06-25 2010-12-30 Amgen Inc. Heterocyclic compounds and their uses
WO2012037204A1 (en) * 2010-09-14 2012-03-22 Exelixis, Inc. Inhibitors of pi3k-delta and methods of their use and manufacture
WO2012061696A1 (en) * 2010-11-04 2012-05-10 Amgen Inc. 5 -cyano-4, 6 -diaminopyrimidine or 6 -aminopurine derivatives as pi3k- delta inhibitors
US20130267524A1 (en) 2012-04-04 2013-10-10 Amgen Inc. Heterocyclic compounds and their uses
US20160016915A1 (en) * 2012-12-21 2016-01-21 Gilead Calistoga Llc Phosphatidylinositol 3-kinase inhibitors

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
"FDA approves Zydelig for three types of blood cancers", 23 July 2014, FOOD AND DRUG ADMINISTRATION
ALI ET AL., NATURE, vol. 510, 2014, pages 407 - 411
BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1
CANTLY, C., SCIENCE, 2002, pages 1655
CUSHING ET AL.: "Discovery and in Vivo Evaluation of (S)-N-(1-(7-Fluoro-2-(pyridin-2-yl)quinolin-3- yl)ethyl)-9H-purin-6-amine (AMG319) and Related PI3K delta inhibitors for Inflammation and Autoimmune Disease", JOURNAL OF MEDICINAL CHEMISTRY, vol. 58, 3 December 2014 (2014-12-03), XP055195514 *
CUSHING, T. ET AL., J MED CHEM., vol. 58, 2015, pages 480
DI PAOLO, G. ET AL., NATURE, vol. 443, 2006, pages 651
HAWKINS, P. T. ET AL., BIOCHEM. SOC. TRANS., vol. 34, 2006, pages 647
IVAN ANGULO ET AL., SCIENCE, vol. 342, 2013, pages 866
J. MED. CHEM., vol. 58, 2015, pages 480 - 511
KNIGHT, Z. ET AL., CELL, vol. 125, 2006, pages 733
PARKER, P. J. ET AL., BIOCHEM. SOC. TRANS., vol. 32, 2004, pages 893
SCHAEFFER, E. M. ET AL., CURR. OPIN. IMMNUNOL., vol. 12, 2000, pages 282
See also references of EP3400227A4

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10792283B2 (en) 2016-03-05 2020-10-06 Hangzhou Zhengxiang Pharmaceuticals Co., Ltd. Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
US11446301B2 (en) 2016-03-05 2022-09-20 Nanjing Zhengxiang Pharmaceuticals Co., Ltd. Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Also Published As

Publication number Publication date
JP7201992B2 (en) 2023-01-11
EP3400227A1 (en) 2018-11-14
KR20210059033A (en) 2021-05-24
CA3124815A1 (en) 2017-09-14
US20210046075A1 (en) 2021-02-18
KR20180104160A (en) 2018-09-19
EP3400227A4 (en) 2019-06-19
CA3013490A1 (en) 2017-09-14
CN108290898B (en) 2020-09-08
EP3400227B1 (en) 2021-03-31
ES2865424T3 (en) 2021-10-15
CA3013490C (en) 2021-09-14
KR102256497B1 (en) 2021-05-26
US10792283B2 (en) 2020-10-06
CN112250665A (en) 2021-01-22
US20190365752A1 (en) 2019-12-05
CN108290898A (en) 2018-07-17
US11446301B2 (en) 2022-09-20
JP2019511452A (en) 2019-04-25

Similar Documents

Publication Publication Date Title
US10167282B2 (en) 1-(5-tert-butyl-2-aryl-pyrazol-3-yl)-3-[2-fluoro-4-[(3-oxo-4H-pyrido [2, 3-B]pyrazin- 8-yl)oxy]phenyl]urea derivatives as RAF inhibitors for the treatment of cancer
US11446301B2 (en) Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
EP3119765B1 (en) Heteroaryl syk inhibitors
JP6868011B2 (en) Pyrazolyl-substituted heteroaryls and their use as pharmaceuticals
US10611777B2 (en) Imidazopyridazine compounds and their use
US20230227446A9 (en) Phosphatidylinositol 3-kinase inhibitors
WO2018090939A1 (en) 8,9-dihydroimidazole[1,2-a]pyrimido[5,4-e]pyrimidine-5(6h)-ketone compound
JP2013542966A (en) Pyrazolopyridines and their use as TYK2 inhibitors and their use
TW201217387A (en) Azabenzothiazole compounds, compositions and methods of use
JP2022507231A (en) 2,3-Dihydro-1H-pyrrolo [3,4-c] pyridin-1-one derivative as an HPK1 inhibitor for the treatment of cancer

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2018533215

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 3013490

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2017763752

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017763752

Country of ref document: EP

Effective date: 20180808

ENP Entry into the national phase

Ref document number: 20187025588

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020187025588

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE